Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 118(18): 4817-28, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21849486

ABSTRACT

We report the findings from the first 10 patients with chemotherapy-refractory chronic lymphocytic leukemia (CLL) or relapsed B-cell acute lymphoblastic leukemia (ALL) we have enrolled for treatment with autologous T cells modified to express 19-28z, a second-generation chimeric antigen (Ag) receptor specific to the B-cell lineage Ag CD19. Eight of the 9 treated patients tolerated 19-28z(+) T-cell infusions well. Three of 4 evaluable patients with bulky CLL who received prior conditioning with cyclophosphamide exhibited either a significant reduction or a mixed response in lymphadenopathy without concomitant development of B-cell aplasia. In contrast, one patient with relapsed ALL who was treated in remission with a similar T-cell dose developed a predicted B-cell aplasia. The short-term persistence of infused T cells was enhanced by prior cyclophosphamide administration and inversely proportional to the peripheral blood tumor burden. Further analyses showed rapid trafficking of modified T cells to tumor and retained ex vivo cytotoxic potential of CD19-targeted T cells retrieved 8 days after infusion. We conclude that this adoptive T-cell approach is promising and more likely to show clinical benefit in the setting of prior conditioning chemotherapy and low tumor burden or minimal residual disease. These studies are registered at www.clinicaltrials.org as #NCT00466531 (CLL protocol) and #NCT01044069 (B-ALL protocol).


Subject(s)
Antigens, CD19/immunology , Graft Survival , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/therapy , T-Lymphocytes/transplantation , Adult , Aged , Antigens, CD19/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/physiology , Female , Graft Survival/physiology , Humans , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/immunology , Male , Middle Aged , Recurrence , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Transplantation, Autologous , Treatment Failure
2.
J Immunother ; 32(2): 169-80, 2009.
Article in English | MEDLINE | ID: mdl-19238016

ABSTRACT

On the basis of promising preclinical data demonstrating the eradication of systemic B-cell malignancies by CD19-targeted T lymphocytes in vivo in severe combined immunodeficient-beige mouse models, we are launching phase I clinical trials in patients with chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia. We present here the validation of the bioprocess which we developed for the production and expansion of clinical grade autologous T cells derived from patients with CLL. We demonstrate that T cells genetically modified with a replication-defective gammaretroviral vector derived from the Moloney murine leukemia virus encoding a chimeric antigen receptor (CAR) targeted to CD19 (1928z) can be expanded with Dynabeads CD3/CD28. This bioprocess allows us to generate clinical doses of 1928z+ T cells in approximately 2 to 3 weeks in a large-scale semiclosed culture system using the Wave Bioreactor. These 1928z+ T cells remain biologically functional not only in vitro but also in severe combined immunodeficient-beige mice bearing disseminated tumors. The validation requirements in terms of T-cell expansion, T-cell transduction with the 1928z CAR, biologic activity, quality control testing, and release criteria were met for all 4 validation runs using apheresis products from patients with CLL. Additionally, after expansion of the T cells, the diversity of the skewed Vbeta T-cell receptor repertoire was significantly restored. This validated process will be used in phase I clinical trials in patients with chemorefractory CLL and in patients with relapsed acute lymphoblastic leukemia. It can also be adapted for other clinical trials involving the expansion and transduction of patient or donor T cells using any CAR or T-cell receptor.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, Antigen/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , Bioreactors , Cell Culture Techniques , Clinical Trials as Topic , Genetic Engineering , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Mice , Receptors, Antigen/immunology , T-Lymphocytes, Cytotoxic/transplantation , Transduction, Genetic
3.
Vaccine ; 25(27): 5013-24, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17537555

ABSTRACT

The use of plasmid DNA as vaccines for the treatment of cancer and infectious diseases is on the rise. In order to facilitate the manufacture of clinical-grade plasmid DNA for Phase I clinical trials, we developed a process whereby >200 mg plasmid could be produced in a single production run under Good Manufacturing Practices. A dedicated cleanroom (Class 10,000 with Class 100 biosafety cabinet) is utilized for production of the bacterial cell bank, fermentation, harvest/lysis of the biomass, and downstream purification. Fermentation requires three 16-18 h runs (approximately 12 L each) in shaker-flasks, yielding approximately 60 g bacterial paste following batch centrifugation. The biomass is alkaline-lysed, pooled, and the resulting flocculent precipitate is separated by a novel vacuum step, followed by depth-filtration. Downstream processing includes anion-exchange chromatography, utilizing Qiagen silica-based resin, and precipitation with isopropanol. Following precipitation, the DNA is harvested by centrifugation, dried, formulated, and sterile-filtered using a Sartorius Sartobran 150 filter prior to Final-Filling. All processing steps utilize sterilized, single-use components. This process results in a product manufactured according to regulatory guidelines. The plasmid DNA is sterile with >or=95% supercoiled DNA, an A260/A280 ratio>or=1.9, undetectable or extremely low residual endotoxin, RNA, genomic DNA, protein, and antibiotic. Residual solvent levels are negligible. The product yields the predicted profile upon restriction-enzyme digestion, is biologically active upon transfection and remains stable for several years at -20 degrees C. We have therefore developed a reproducible and cost effective process to manufacture clinical-grade plasmid DNA. This process can be adapted by other academic centers for human or large animal clinical trials.


Subject(s)
Clinical Trials, Phase I as Topic/standards , DNA/biosynthesis , Drug Industry , Plasmids/genetics , Vaccines, DNA/biosynthesis , Animals , Cellulose/analogs & derivatives , Chromatography, Ion Exchange , Chromosome Mapping , Culture Media , DNA/analysis , Electrophoresis, Polyacrylamide Gel , Fermentation , Humans , Indicators and Reagents , Vaccines, DNA/analysis
4.
Hum Gene Ther ; 17(7): 780-94, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16839276

ABSTRACT

The successful transduction and engraftment of human mobilized peripheral blood (MBP) CD34(+) cells are determined to a large extent by the ex vivo cell-processing conditions. In preparation for upcoming clinical trials, we investigated essential culture parameters and devised a short and efficient gammaretroviral transduction protocol entailing minimal manipulation of MBP CD34(+) cells. The engraftment potential and in vivo transgene expression in the progeny of repopulating CD34(+) cells were measured to assess the functionality of CD34(+) cells transduced under these conditions. Using a competitive in vivo repopulation assay in nonobese diabetic/severe combined immunodeficient mice, we demonstrate equivalent engraftment of CD34(+) cells transduced under serum-free conditions as compared with CD34(+) cells cultured with serum. We also took advantage of this in vivo model to demonstrate that ex vivo manipulation of CD34(+) cells can be shortened to 60 hr, using 36 hr of prestimulation and two cycles of transduction 12 hr apart. These minimally manipulated CD34(+) cells engraft in a manner similar to cells transduced under longer protocols and the vector-encoded transgene is expressed at the same frequency in cells derived from repopulating CD34(+) cells in vivo. We have thus developed a short and efficient human MBP CD34(+) transduction protocol under serum-free conditions that is suitable and broadly applicable for phase I clinical trials.


Subject(s)
Antigens, CD34/analysis , Blood Cells/transplantation , Gammaretrovirus/genetics , Genetic Therapy/methods , Animals , Blood Cells/chemistry , Cattle , Cell Culture Techniques , Female , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred Strains , Serum/cytology , Stem Cell Factor/genetics , Tetrahydrofolate Dehydrogenase/genetics , Transduction, Genetic , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...